Joseph M. Salamoun
University of Pittsburgh
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Joseph M. Salamoun.
Bioorganic & Medicinal Chemistry Letters | 2015
Wei Qian; Joseph M. Salamoun; Jingnan Wang; Vera Roginskaya; Bennett Van Houten; Peter Wipf
The effective management of tumors resistant to platinum drugs-based anticancer therapies is a critical challenge in current clinical practices. The proapoptotic Bcl-2 family proteins Bax and Bak are essential for cisplatin-induced apoptosis. Unfortunately, Bax and its related upstream endogenous apoptotic signaling pathways are often dysregulated in cancer cells. Strategies that are able to bypass Bax- and Bak-dependent apoptotic pathways will thus provide opportunities to overcome platinum drug resistance. We have identified the thioxodihydroquinazolinone mdivi-1 as a member of a novel class of small molecules that are able to induce Bax- and Bak-independent mitochondrial outer membrane permeabilization when combined with cisplatin, thereby efficiently triggering apoptosis in platinum-resistant tumor cells. In the present structure activity relationship (SAR) study of a computationally selected library of mdivi-1 related small molecules, we established a pharmacophore model that can lead to the enhancement of platinum drug efficacy and Bax/Bak-independent mitochondrial apoptosis. Specifically, we found that a thiourea function is necessary but not sufficient for the synergism of this class of thioxodihydroquinazolinones with cisplatin. We were also able to identify more potent mdivi-1 analogs through this SAR study, which will guide future designs with the goal to develop novel combination regimens for the treatment of platinum- and multidrug-resistant tumors.
PLOS ONE | 2015
Manuj Tandon; Joseph M. Salamoun; Evan J. Carder; Elisa Farber; Shuping Xu; Fan Deng; Hua Tang; Peter Wipf; Q. Jane Wang
Protein kinase D (PKD) has been implicated in many aspects of tumorigenesis and progression, and is an emerging molecular target for the development of anticancer therapy. Despite recent advancement in the development of potent and selective PKD small molecule inhibitors, the availability of in vivo active PKD inhibitors remains sparse. In this study, we describe the discovery of a novel PKD small molecule inhibitor, SD-208, from a targeted kinase inhibitor library screen, and the synthesis of a series of analogs to probe the structure-activity relationship (SAR) vs. PKD1. SD-208 displayed a narrow SAR profile, was an ATP-competitive pan-PKD inhibitor with low nanomolar potency and was cell active. Targeted inhibition of PKD by SD-208 resulted in potent inhibition of cell proliferation, an effect that could be reversed by overexpressed PKD1 or PKD3. SD-208 also blocked prostate cancer cell survival and invasion, and arrested cells in the G2/M phase of the cell cycle. Mechanistically, SD-208-induced G2/M arrest was accompanied by an increase in levels of p21 in DU145 and PC3 cells as well as elevated phosphorylation of Cdc2 and Cdc25C in DU145 cells. Most importantly, SD-208 given orally for 24 days significantly abrogated the growth of PC3 subcutaneous tumor xenografts in nude mice, which was accompanied by reduced proliferation and increased apoptosis and decreased expression of PKD biomarkers including survivin and Bcl-xL. Our study has identified SD-208 as a novel efficacious PKD small molecule inhibitor, demonstrating the therapeutic potential of targeted inhibition of PKD for prostate cancer treatment.
Organic Letters | 2014
Joseph M. Salamoun; Shelby M. Anderson; James C. Burnett; Rick Gussio; Peter Wipf
Two complementary approaches for the preparation of linked 5-membered heterocycles were developed. The Pd-catalyzed Suzuki–Miyaura cross-coupling with halogenated furan, thiophene, and selenophene led to higher overall yields, but C,H-bond activation was a more efficient strategy for the coupling at C(2) of oxazoles. Potency and selectivity of the final hydroxymethyl products in renal (A498), lung (NCI-H226), kidney (CAKI-1), and breast (MDA-MB-468, MCF7) carcinoma cell lines were determined.
Oncotarget | 2017
Kelley E. McQueeney; Joseph M. Salamoun; James C. Burnett; Nektarios Barabutis; Paula Pekic; Sophie Lewandowski; Danielle C. Llaneza; Robert Cornelison; Yunpeng Bai; Zhong Yin Zhang; John D. Catravas; Charles N. Landen; Peter Wipf; John S. Lazo; Elizabeth R. Sharlow
Overexpression of protein tyrosine phosphatase PTP4A oncoproteins is common in many human cancers and is associated with poor patient prognosis and survival. We observed elevated levels of PTP4A3 phosphatase in 79% of human ovarian tumor samples, with significant overexpression in tumor endothelium and pericytes. Furthermore, PTP4A phosphatases appear to regulate several key malignant processes, such as invasion, migration, and angiogenesis, suggesting a pivotal regulatory role in cancer and endothelial signaling pathways. While phosphatases are attractive therapeutic targets, they have been poorly investigated because of a lack of potent and selective chemical probes. In this study, we disclose that a potent, selective, reversible, and noncompetitive PTP4A inhibitor, JMS-053, markedly enhanced microvascular barrier function after exposure of endothelial cells to vascular endothelial growth factor or lipopolysaccharide. JMS-053 also blocked the concomitant increase in RhoA activation and loss of Rac1. In human ovarian cancer cells, JMS-053 impeded migration, disrupted spheroid growth, and decreased RhoA activity. Importantly, JMS-053 displayed anticancer activity in a murine xenograft model of drug resistant human ovarian cancer. These data demonstrate that PTP4A phosphatases can be targeted in both endothelial and ovarian cancer cells, and confirm that RhoA signaling cascades are regulated by the PTP4A family.
Oncotarget | 2017
Jing Ma; Joseph M. Salamoun; Peter Wipf; Robert P. Edwards; Bennett Van Houten; Wei Qian
Cancer stem cell-like cells (CSC-LCs) contribute to drug resistance and recurrence of ovarian cancer. Strategies that can eradicate CSC-LCs are expected to substantially improve the outcome of ovarian cancer treatment. We have previously identified a class of thioxodihydroquinazolinone small molecules, which have strong synergistic antitumor activity with platinum drugs, the standard chemotherapeutic agents for ovarian cancer treatment. In the current study, using the activity of aldehyde dehydrogenase (ALDH) as a marker of CSC-LCs, we demonstrated that the combination of thioxodihydroquinazolinone compound 19 with cisplatin is able to diminish ALDH-high CSC-LC populations in both platinum-resistant ovarian cancer cell lines and primary ovarian cancer cells from metastatic ascites of a cisplatin-resistant patient. Compound 19 enhanced the accumulation of intracellular cisplatin in ALDH-high ovarian CSC-LCs. The combination of compound 19 with cisplatin was also able to reduce the sphere-forming capability of cisplatin-resistant ovarian cancer cells. Using a spheroid-based in vitro metastasis model of ovarian cancer, we demonstrated that the co-administration of compound 19 with cisplatin prevents ovarian cancer spheroid cells from attaching to substratum and spreading. In a cisplatin-resistant in vivo intraperitoneal xenograft mouse model, the combination of compound 19 with cisplatin significantly reduced tumor burden, as compared to cisplatin alone. Taken together, our study demonstrated that thioxodihydroquinazolinones represent a new class of agents that in combination with cisplatin are capable of eliminating CSC-LCs in ovarian cancer. Further development of thioxodihydroquinazolinone small molecules may yield a more effective treatment for cisplatin-resistant metastatic ovarian cancer.
Journal of Medicinal Chemistry | 2016
Joseph M. Salamoun; Peter Wipf
Despite their extensive involvement in signaling pathways and disease pathologies, targeting protein phosphatases remains an underexplored opportunity in drug discovery. Selective intracellular regulation of phosphatases with small molecule inhibitors has been an unmet challenge. However, recent progress in the development of allosteric modulators encourages renewed efforts to exploit their potential as therapeutic targets.
The FASEB Journal | 2018
Kelley E. McQueeney; Joseph M. Salamoun; Jennifer Ahn; Paula Pekic; Isabella K. Blanco; Heather Struckman; Elizabeth R. Sharlow; Peter Wipf; John S. Lazo
Dysregulation of the tightly controlled protein phosphorylation networks that govern cellular behavior causes cancer. The membrane‐associated, intracellular protein tyrosine phosphatase PTP4A3 is overexpressed in human colorectal cancer and contributes to cell migration and invasion. To interrogate further the role of PTP4A3 in colorectal cancer cell migration and invasion, we deleted the Ptp4a3 gene from murine colorectal tumor cells. The resulting PTP4A3−/− cells exhibited impaired colony formation, spheroid formation, migration, and adherence compared with the paired PTP4A3fl/fl cells. We replicated these phenotypic changes using the new small‐molecule, allosteric PTP4A3 inhibitor JMS‐053. A related structure, JMS‐038, which lacked phosphatase inhibition, displayed no cellular activity. Reduction in cell viability and colony formation by JMS‐053 occurred in both mouse and human colorectal cell lines and required PTP4A3 expression. Ptp4a3 deletion increased the expression of extracellular matrix (ECM) and adhesion genes, including the tumor suppressor Emilin 1. JMS‐ 053 also increased Emilin 1 gene expression. Moreover, The Cancer Genome Atlas genomic database revealed human colorectal tumors with high Ptp4a3 expression had low Emilin 1 expression. These chemical and biologic reagents reveal a previously unknown communication between the intracellular PTP4A3 phosphatase and the ECM and support efforts to pharmacologically target PTP4A3.—McQueeney, K. E., Salamoun, J. M., Ahn, J. G., Pekic, P., Blanco, I. K., Struckman, H. L., Sharlow, E. R., Wipf, P., Lazo, J. S. A chemical genetics approach identifies PTP4A3 as a regulator of colon cancer cell adhesion. FASEB J. 32, 5661–5673 (2018). www.fasebj.org
Cancer Research | 2017
John S. Lazo; Paula Pekic; Alex Cheung; Kelley E. McQueeney; Joseph M. Salamoun; Peter Wipf; Charles N. Landen; Elizabeth R. Sharlow
PTP4A3 is a highly attractive molecular target for ovarian cancer (OvCa). Elevated levels of PTP4A3 mRNA and protein in human ovarian tumors correlate with disease progression, poor prognosis and poor survival. Genetic depletion of PTP4A3 in OvCa cell lines diminishes their ability to migrate and reduces their in vivo tumorigenicity while PTP4A3 overexpression increases tumor cell migration, invasion, and dissemination. Together, these data suggest PTP4A3 is a novel molecular target for OvCa; however, the lack of potent and selective PTP4A3 small molecule inhibitors has hindered PTP4A3’s definitive validation in OvCa and in other cancers. To drive the pharmacological validation of PTP4A3, we developed JMS-631-053 (7-imino-2-phenylthieno[3,2-c]pyridine-4,6(5H,7H)-dione), a potent (K i =3 nM in vitro), specific, noncompetitive, PTP4A3 inhibitor. JMS-631-053 inhibited cellular migration, invasion, and colony formation in soft agar. Potent OvCa cell-based effects were observed by profiling JMS-631-053 against a panel of 8 OvCa cell lines using a 2D drug susceptibility assay, with EC 50 values as low as 600 nM. Likewise, JMS-631-053 killed OvCa 3D spheroids, including those derived from high grade serous OvCa cell lines, with EC 50 values as low as 300 nM. The OvCa drug resistant cell lines (i.e., A2780CP20 and HeyA8MDR) retained responsiveness to JMS-631-053 (using 2D and 3D culturing conditions) suggesting that inhibition of PTP4A3 may be a viable therapeutic strategy for chemoresistant OvCa. Preliminary data also suggests that JMS-631-053 synergizes with cisplatin when used in combination studies. Hence, JMS-631-053 will be a valuable chemical tool for further validation of PTP4A3 as an OvCa target as well as provide potential leads for future drug discovery. Citation Format: John S. Lazo, Paula Pekic, Alex Cheung, Kelley McQueeney, Joseph Salamoun, Peter Wipf, Charles N. Landen, Elizabeth R. Sharlow. Targeting PTP4A3 phosphatase in ovarian cancer with the potent noncompetitive inhibitor JMS-631-053 [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 3210. doi:10.1158/1538-7445.AM2017-3210
Cancer Research | 2017
Kelley E. McQueeney; Joseph M. Salamoun; Isabella K. Blanco; Paula Pekic; Jennifer Ahn; Elizabeth R. Sharlow; Peter Wipf; John S. Lazo
Aberrant regulation of protein phosphorylation is an exceedingly common driver of human cancers. It is notable that we understand much less about the role of protein tyrosine phosphatases in human malignancies compared to tyrosine kinases. The membrane-associated, intracellular, protein tyrosine PTP4A3 is highly overexpressed in multiple tumor types including colorectal cancer and has been associated with tumor metastases. We have, therefore, investigated the role of PTP4A3 in colon cancer migration and invasion. The Ptp4a3 gene was expunged from colon tumor cells derived from Ptp4a3 flox/flox mice and the resulting cells exhibited impaired migration, invasion, and colony formation compared to the wildtype isogenic cells. We characterized a potent, selective, and noncompetitive small molecule inhibitor of PTP4A3, JMS-631-053, which also disrupted colon cancer cell migration, invasion, and colony formation. PTP4A3 deletion increased the expression of extracellular matrix and adhesion genes, including the tumor suppressor Emilin 1. Expression of these extracellular matrix genes is mutually exclusive with PTP4A3 expression in tumors derived from patients with colorectal cancer. These chemical and biological reagents reveal a previously unknown communication between the intracellular PTP4A3 phosphatase and the extracellular matrix and support continued efforts to pharmacologically target PTP4A3 for cancer therapy. Citation Format: Kelley E. McQueeney, Joseph M. Salamoun, Isabella K. Blanco, Paula Pekic, Jennifer Ahn, Elizabeth R. Sharlow, Peter Wipf, John S. Lazo. Coordinated chemical-genetics approach identifies PTP4A3-mediated regulation of colon cancer cell migration and extracellular matrix interactions [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 1220. doi:10.1158/1538-7445.AM2017-1220
Clinical Cancer Research | 2016
Elizabeth R. Sharlow; Joseph M. Salamoun; Kelley E. McQueeney; Sophie Lewandowski; Jennifer L. Bryant; Alex Cheung; Paula Pekic; Charles N. Landen; Peter Wipf; John S. Lazo
PTP4A3 (also known as PRL-3) is a unique protein tyrosine phosphatase that is highly expressed in human ovarian cancer patient samples. Genetic knockdown studies implicate PTP4A3 in ovarian cancer tumorigenesis and the maintenance of the malignant phenotype. Interestingly, cBioportal data mining reveals that PTP4A3 is one of the most highly amplified genes in ovarian cancer, implicating PTP4A3 as a novel, and to date, largely underexplored potential molecular therapeutic target. Unfortunately, the lack of potent and selective PTP4A3 inhibitors impedes the validation of PTP4A3 as a drug target for ovarian cancer. To drive target validation studies and complement our ongoing cell-based genetic PTP4A3 knockdown strategies, we have developed novel PTP4A3 small molecule inhibitors to delineate its mechanistic role in ovarian cancer and therapeutic potential as a drug target. We resynthesized the most potent published PTP4A3 inhibitor (thienopyridone or JMS-631-050). Concurrently, we discovered a new, more active thienopyridone analog, JMS-631-053. Additionally, we identified a structurally related inactive analog, JMS-557-038, which could be used as a pharmacologically powerful control. JMS-631-050 and JMS-631-053 inhibited recombinant PTP4A3 in vitro phosphatase activity with IC50 values of 138 and 7 nM, respectively. We then profiled the impact of JMS-631-050, JMS-631-053 and JMS-557-038 on cell adhesion-based survival using a panel of eight ovarian cancer cell lines, which included a representative high serous grade ovarian cancer cell line (i.e., OVCAR4) and several drug sensitive/resistant cell line pairs. In all ovarian cancer cells lines tested, both JMS-631-050 and JMS-631-053 inhibited cell adhesion-mediated survival. However, JMS-631-053 displayed more potent cell-based effects than JMS-631-050 in all cell lines evaluated. A2780 cells were the most sensitive to JMS-631-053 effects (EC50=0.60±0.2 μM). Significantly, OVCAR4 cells were also highly sensitive to JMS-631-053 with an EC50=1.5±0.3 μM. Both JMS-631-050 and JMS-631-053 maintained cell-based activity when evaluated in three dimensional cell culture models. As expected the PTP4A3 inactive analog, JMS-557-038 had no effect on cell-adhesion-mediated survival (EC50s>50 μM) in any of the ovarian cancer cell lines tested. Thus, JMS-631-050 and JMS-631-053 are attractive chemotypes for further evaluation as single agents for ovarian cancer. Moreover, as the drug resistant cell lines A2780CP20 and HeyA8MDR retained sensitivity to JMS-631-050 and JMS-631-053, these chemotypes are under evaluation in combination studies using existing ovarian cancer therapies. Hence, JMS-631-050 and JMS-631-053 will be valuable reagents for further validation of PTP4A3 as an ovarian cancer target as well as potential leads for future drug dis. Citation Format: Elizabeth R. Sharlow, Joseph M. Salamoun, Kelley E. McQueeney, Sophie Lewandowski, Jennifer Bryant, Alex Cheung, Paula Pekic, Charles N. Landen, Jr., Peter Wipf, John S. Lazo. Profiling potent, novel protein tyrosine phosphatase 4A3 small molecule inhibitors for ovarian cancer. [abstract]. In: Proceedings of the AACR Special Conference on Advances in Ovarian Cancer Research: Exploiting Vulnerabilities; Oct 17-20, 2015; Orlando, FL. Philadelphia (PA): AACR; Clin Cancer Res 2016;22(2 Suppl):Abstract nr A83.